Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Large-scale genetic study in East Asians identifies six new loci associated with colorectal cancer risk

Abstract

Known genetic loci explain only a small proportion of the familial relative risk of colorectal cancer (CRC). We conducted a genome-wide association study of CRC in East Asians with 14,963 cases and 31,945 controls and identified 6 new loci associated with CRC risk (P = 3.42 × 10−8 to 9.22 × 10−21) at 10q22.3, 10q25.2, 11q12.2, 12p13.31, 17p13.3 and 19q13.2. Two of these loci map to genes (TCF7L2 and TGFB1) with established roles in colorectal tumorigenesis. Four other loci are located in or near genes involved in transcriptional regulation (ZMIZ1), genome maintenance (FEN1), fatty acid metabolism (FADS1 and FADS2), cancer cell motility and metastasis (CD9), and cell growth and differentiation (NXN). We also found suggestive evidence for three additional loci associated with CRC risk near genome-wide significance at 8q24.11, 10q21.1 and 10q24.2. Furthermore, we replicated 22 previously reported CRC-associated loci. Our study provides insights into the genetic basis of CRC and suggests the involvement of new biological pathways.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Forest plots for risk-associated variants in the six newly identified loci.
Figure 2: Association of selected risk variants identified in this study with gene expression in colon tumor tissue.

Similar content being viewed by others

References

  1. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    PubMed  Google Scholar 

  2. Lichtenstein, P. et al. Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark, and Finland. N. Engl. J. Med. 343, 78–85 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. de la Chapelle, A. Genetic predisposition to colorectal cancer. Nat. Rev. Cancer 4, 769–780 (2004).

    Article  CAS  PubMed  Google Scholar 

  4. Aaltonen, L., Johns, L., Jarvinen, H., Mecklin, J.P. & Houlston, R. Explaining the familial colorectal cancer risk associated with mismatch repair (MMR)-deficient and MMR-stable tumors. Clin. Cancer Res. 13, 356–361 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Ma, X., Zhang, B. & Zheng, W. Genetic variants associated with colorectal cancer risk: comprehensive research synopsis, meta-analysis, and epidemiological evidence. Gut 63, 326–336 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Palles, C. et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat. Genet. 45, 136–144 (2013).

    Article  CAS  PubMed  Google Scholar 

  7. Zanke, B.W. et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on chromosome 8q24. Nat. Genet. 39, 989–994 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Tomlinson, I. et al. A genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat. Genet. 39, 984–988 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Broderick, P. et al. A genome-wide association study shows that common alleles of SMAD7 influence colorectal cancer risk. Nat. Genet. 39, 1315–1317 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Jaeger, E. et al. Common genetic variants at the CRAC1 (HMPS) locus on chromosome 15q13.3 influence colorectal cancer risk. Nat. Genet. 40, 26–28 (2008).

    Article  CAS  PubMed  Google Scholar 

  11. Tenesa, A. et al. Genome-wide association scan identifies a colorectal cancer susceptibility locus on 11q23 and replicates risk loci at 8q24 and 18q21. Nat. Genet. 40, 631–637 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Tomlinson, I.P. et al. A genome-wide association study identifies colorectal cancer susceptibility loci on chromosomes 10p14 and 8q23.3. Nat. Genet. 40, 623–630 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Houlston, R.S. et al. Meta-analysis of genome-wide association data identifies four new susceptibility loci for colorectal cancer. Nat. Genet. 40, 1426–1435 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Houlston, R.S. et al. Meta-analysis of three genome-wide association studies identifies susceptibility loci for colorectal cancer at 1q41, 3q26.2, 12q13.13 and 20q13.33. Nat. Genet. 42, 973–977 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tomlinson, I.P. et al. Multiple common susceptibility variants near BMP pathway loci GREM1, BMP4, and BMP2 explain part of the missing heritability of colorectal cancer. PLoS Genet. 7, e1002105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dunlop, M.G. et al. Common variation near CDKN1A, POLD3 and SHROOM2 influences colorectal cancer risk. Nat. Genet. 44, 770–776 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Peters, U. et al. Identification of genetic susceptibility loci for colorectal tumors in a genome-wide meta-analysis. Gastroenterology 144, 799–807 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Jia, W.H. et al. Genome-wide association analyses in East Asians identify new susceptibility loci for colorectal cancer. Nat. Genet. 45, 191–196 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, B. et al. Genome-wide association study identifies a new SMAD7 risk variant associated with colorectal cancer risk in East Asians. Int. J. Cancer 10.1002/ijc.28733 (21 January 2014).

  20. Cui, R. et al. Common variant in 6q26-q27 is associated with distal colon cancer in an Asian population. Gut 60, 799–805 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Figueiredo, J.C. et al. Genotype-environment interactions in microsatellite stable/microsatellite instability-low colorectal cancer: results from a genome-wide association study. Cancer Epidemiol. Biomarkers Prev. 20, 758–766 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Abecasis, G.R. et al. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Frazer, K.A. et al. A second generation human haplotype map of over 3.1 million SNPs. Nature 449, 851–861 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  25. Westra, H.J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Degner, J.F. et al. DNase I sensitivity QTLs are a major determinant of human expression variation. Nature 482, 390–394 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

  28. Grundberg, E. et al. Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nat. Genet. 44, 1084–1089 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Forbes, S.A. et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 39, D945–D950 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Cancer Genome Atlas Network. Comprehensive molecular characterization of human colon and rectal cancer. Nature 487, 330–337 (2012).

  31. Kapushesky, M. et al. Gene Expression Atlas update—a value-added database of microarray and sequencing-based functional genomics experiments. Nucleic Acids Res. 40, D1077–D1081 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Tang, W. et al. A genome-wide RNAi screen for Wnt/β-catenin pathway components identifies unexpected roles for TCF transcription factors in cancer. Proc. Natl. Acad. Sci. USA 105, 9697–9702 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Angus-Hill, M.L., Elbert, K.M., Hidalgo, J. & Capecchi, M.R. T-cell factor 4 functions as a tumor suppressor whose disruption modulates colon cell proliferation and tumorigenesis. Proc. Natl. Acad. Sci. USA 108, 4914–4919 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Bass, A.J. et al. Genomic sequencing of colorectal adenocarcinomas identifies a recurrent VTI1A-TCF7L2 fusion. Nat. Genet. 43, 964–968 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Grainger, D.J. et al. Genetic control of the circulating concentration of transforming growth factor type β1. Hum. Mol. Genet. 8, 93–97 (1999).

    Article  CAS  PubMed  Google Scholar 

  36. Kumar, P., Henikoff, S. & Ng, P.C. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 4, 1073–1081 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Adzhubei, I.A. et al. A method and server for predicting damaging missense mutations. Nat. Methods 7, 248–249 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dunning, A.M. et al. A transforming growth factor β1 signal peptide variant increases secretion in vitro and is associated with increased incidence of invasive breast cancer. Cancer Res. 63, 2610–2615 (2003).

    CAS  PubMed  Google Scholar 

  39. Suthanthiran, M. et al. Transforming growth factor-β1 hyperexpression in African-American hypertensives: a novel mediator of hypertension and/or target organ damage. Proc. Natl. Acad. Sci. USA 97, 3479–3484 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Yamada, Y. et al. Association of a polymorphism of the transforming growth factor-β1 gene with genetic susceptibility to osteoporosis in postmenopausal Japanese women. J. Bone Miner. Res. 13, 1569–1576 (1998).

    Article  CAS  PubMed  Google Scholar 

  41. Markowitz, S.D. & Bertagnolli, M.M. Molecular origins of cancer: molecular basis of colorectal cancer. N. Engl. J. Med. 361, 2449–2460 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Howe, J.R. et al. Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science 280, 1086–1088 (1998).

    Article  CAS  PubMed  Google Scholar 

  43. Valle, L. et al. Germline allele-specific expression of TGFBR1 confers an increased risk of colorectal cancer. Science 321, 1361–1365 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Liu, L. et al. Functional FEN1 genetic variants contribute to risk of hepatocellular carcinoma, esophageal cancer, gastric cancer and colorectal cancer. Carcinogenesis 33, 119–123 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Xu, Z. & Taylor, J.A. SNPinfo: integrating GWAS and candidate gene information into functional SNP selection for genetic association studies. Nucleic Acids Res. 37, W600–W605 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zheng, L. et al. Functional regulation of FEN1 nuclease and its link to cancer. Nucleic Acids Res. 39, 781–794 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Zheng, L. et al. Fen1 mutations result in autoimmunity, chronic inflammation and cancers. Nat. Med. 13, 812–819 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Kucherlapati, M. et al. Haploinsufficiency of Flap endonuclease (Fen1) leads to rapid tumor progression. Proc. Natl. Acad. Sci. USA 99, 9924–9929 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Schaeffer, L. et al. Common genetic variants of the FADS1-FADS2 gene cluster and their reconstructed haplotypes are associated with the fatty acid composition in phospholipids. Hum. Mol. Genet. 15, 1745–1756 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Castellone, M.D., Teramoto, H., Williams, B.O., Druey, K.M. & Gutkind, J.S. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin–β-catenin signaling axis. Science 310, 1504–1510 (2005).

    Article  CAS  PubMed  Google Scholar 

  51. Cai, Q. et al. Prospective study of urinary prostaglandin E2 metabolite and colorectal cancer risk. J. Clin. Oncol. 24, 5010–5016 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Rogers, L.M., Riordan, J.D., Swick, B.L., Meyerholz, D.K. & Dupuy, A.J. Ectopic expression of Zmiz1 induces cutaneous squamous cell malignancies in a mouse model of cancer. J. Invest. Dermatol. 133, 1863–1869 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Turnbull, C. et al. Genome-wide association study identifies five new breast cancer susceptibility loci. Nat. Genet. 42, 504–507 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ovalle, S. et al. The tetraspanin CD9 inhibits the proliferation and tumorigenicity of human colon carcinoma cells. Int. J. Cancer 121, 2140–2152 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Mori, M. et al. Motility related protein 1 (MRP1/CD9) expression in colon cancer. Clin. Cancer Res. 4, 1507–1510 (1998).

    CAS  PubMed  Google Scholar 

  56. Lee, J.H. et al. Glycoprotein 90K, downregulated in advanced colorectal cancer tissues, interacts with CD9/CD82 and suppresses the Wnt/β-catenin signal via ISGylation of β-catenin. Gut 59, 907–917 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Braumüller, H. et al. T-helper-1-cell cytokines drive cancer into senescence. Nature 494, 361–365 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Wolf, M.J., Seleznik, G.M., Zeller, N. & Heikenwalder, M. The unexpected role of lymphotoxin β receptor signaling in carcinogenesis: from lymphoid tissue formation to liver and prostate cancer development. Oncogene 29, 5006–5018 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Lukashev, M. et al. Targeting the lymphotoxin-β receptor with agonist antibodies as a potential cancer therapy. Cancer Res. 66, 9617–9624 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Funato, Y. & Miki, H. Nucleoredoxin, a novel thioredoxin family member involved in cell growth and differentiation. Antioxid. Redox Signal. 9, 1035–1057 (2007).

    Article  CAS  PubMed  Google Scholar 

  61. Funato, Y., Michiue, T., Asashima, M. & Miki, H. The thioredoxin-related redox-regulating protein nucleoredoxin inhibits Wnt–β-catenin signalling through Dishevelled. Nat. Cell Biol. 8, 501–508 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Michailidou, K. et al. Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat. Genet. 45, 353–361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Eeles, R.A. et al. Identification of 23 new prostate cancer susceptibility loci using the iCOGS custom genotyping array. Nat. Genet. 45, 385–391 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Abnet, C.C. et al. A shared susceptibility locus in PLCE1 at 10q23 for gastric adenocarcinoma and esophageal squamous cell carcinoma. Nat. Genet. 42, 764–767 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Amundadottir, L. et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat. Genet. 41, 986–990 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bei, J.X. et al. A genome-wide association study of nasopharyngeal carcinoma identifies three new susceptibility loci. Nat. Genet. 42, 599–603 (2010).

    Article  CAS  PubMed  Google Scholar 

  67. Nakata, I. et al. Association between the SERPING1 gene and age-related macular degeneration and polypoidal choroidal vasculopathy in Japanese. PLoS ONE 6, e19108 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jee, S.H. et al. Adiponectin concentrations: a genome-wide association study. Am. J. Hum. Genet. 87, 545–552 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zheng, W. et al. Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat. Genet. 41, 324–328 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, Y., Willer, C.J., Ding, J., Scheet, P. & Abecasis, G.R. MaCH: using sequence and genotype data to estimate haplotypes and unobserved genotypes. Genet. Epidemiol. 34, 816–834 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Howie, B., Fuchsberger, C., Stephens, M., Marchini, J. & Abecasis, G.R. Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nat. Genet. 44, 955–959 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  PubMed  Google Scholar 

  73. Freedman, M.L. et al. Assessing the impact of population stratification on genetic association studies. Nat. Genet. 36, 388–393 (2004).

    Article  CAS  PubMed  Google Scholar 

  74. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Willer, C.J., Li, Y. & Abecasis, G.R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lau, J., Ioannidis, J.P. & Schmid, C.H. Quantitative synthesis in systematic reviews. Ann. Intern. Med. 127, 820–826 (1997).

    Article  CAS  PubMed  Google Scholar 

  77. Barrett, J.C., Fry, B., Maller, J. & Daly, M.J. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics 21, 263–265 (2005).

    Article  CAS  PubMed  Google Scholar 

  78. Zheng, W. et al. Common genetic determinants of breast-cancer risk in East Asian women: a collaborative study of 23 637 breast cancer cases and 25 579 controls. Hum. Mol. Genet. 22, 2539–2550 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Johns, L.E. & Houlston, R.S. A systematic review and meta-analysis of familial colorectal cancer risk. Am. J. Gastroenterol. 96, 2992–3003 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Pruim, R.J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Johnson, A.D. et al. SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics 24, 2938–2939 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Ward, L.D. & Kellis, M. HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 40, D930–D934 (2012).

    Article  CAS  PubMed  Google Scholar 

  83. Yan, G. et al. Genome sequencing and comparison of two nonhuman primate animal models, the cynomolgus and Chinese rhesus macaques. Nat. Biotechnol. 29, 1019–1023 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors are solely responsible for the scientific content of this paper. The sponsors of this study had no role in study design, data collection, analysis or interpretation, writing of the report or the decision for submission. We thank all study participants and research staff of all parent studies for their contributions and commitment to this project, R. Courtney for DNA preparation, J. He for data processing and analyses, X. Guo for suggestions on bioinformatics analysis, and M.J. Daly and B.J. Rammer for editing and preparing the manuscript. The work at the Vanderbilt University School of Medicine was supported by US National Institutes of Health (NIH) grants R37CA070867, R01CA082729, R01CA124558, R01CA148667 and R01CA122364, as well as by Ingram Professorship and Research Reward funds from the Vanderbilt University School of Medicine. Studies (grant support) participating in the Asia Colorectal Cancer Consortium include the Shanghai Women's Health Study (US NIH, R37CA070867), the Shanghai Men's Health Study (US NIH, R01CA082729), the Shanghai Breast and Endometrial Cancer Studies (US NIH, R01CA064277 and R01CA092585; contributing only controls), Shanghai Colorectal Cancer Study 3 (US NIH, R37CA070867 and Ingram Professorship funds), the Guangzhou Colorectal Cancer Study (National Key Scientific and Technological Project, 2011ZX09307-001-04; the National Basic Research Program, 2011CB504303, contributing only controls; the Natural Science Foundation of China, 81072383, contributing only controls), the Japan BioBank Colorectal Cancer Study (grant from the Ministry of Education, Culture, Sports, Science and Technology of the Japanese government), the Hwasun Cancer Epidemiology Study–Colon and Rectum Cancer (HCES-CRC; grants from the Korea Center for Disease Control and Prevention and the Jeonnam Regional Cancer Center), the Aichi Colorectal Cancer Study (Grant-in-Aid for Cancer Research, grant for the Third Term Comprehensive Control Research for Cancer and Grants-in-Aid for Scientific Research from the Japanese Ministry of Education, Culture, Sports, Science and Technology, 17015018 and 221S0001), the Korea-NCC (National Cancer Center) Colorectal Cancer Study (Basic Science Research Program through the National Research Foundation of Korea, 2010-0010276; National Cancer Center Korea, 0910220), the Korea-Seoul Colorectal Cancer Study (none reported) and the KCPS-II Colorectal Cancer Study (National R&D Program for Cancer Control, 1220180; Seoul R&D Program, 10526).

We also thank all participants, staff and investigators from the GECCO, CORECT and CCFR consortia for making it possible to present results from populations of European ancestry for the new CRC-associated loci identified among East Asians. GECCO, CORECT and CCFR are directed by U. Peters, S. Gruber and G. Casey, respectively. Complete lists of investigators from the GECCO, CORECT and CCFR consortia are provided below.

Investigators (institution and location) in the GECCO consortium include (in alphabetical order) John A. Baron (Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA), Sonja I. Berndt (Division of Cancer Epidemiology and Genetics, National Cancer Institute, US NIH, Bethesda, Maryland, USA), Stéphane Bezieau (Service de Génétique Médicale, Centre Hospitalier Universitaire (CHU) Nantes, Nantes, France), Hermann Brenner (Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany), Bette J. Caan (Division of Research, Kaiser Permanente Medical Care Program, Oakland, California, USA), Christopher S. Carlson (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, School of Public Health, University of Washington, Seattle, Washington, USA), Graham Casey (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Andrew T. Chan (Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA and Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA), Jenny Chang-Claude (Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany), Stephen J. Chanock (Division of Cancer Epidemiology and Genetics, National Cancer Institute, US NIH, Bethesda, Maryland, USA), David V. Conti (Department of Preventive Medicine, University of Southern California, Los Angeles, California, USA), Keith Curtis (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), David Duggan (Translational Genomics Research Institute, Phoenix, Arizona, USA), Charles S. Fuchs (Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA and Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA), Steven Gallinger (Department of Surgery, Mount Sinai Hospital, Toronto, Ontario, Canada and Samuel Lunenfeld Research Institute, Toronto, Ontario, Canada), Edward L. Giovannucci (Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA, Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA and Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA), Stephen B. Gruber (University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Robert W. Haile (Department of Preventive Medicine, University of Southern California, Los Angeles, California, USA), Tabitha A. Harrison (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Richard B. Hayes (Division of Epidemiology, Department of Environmental Medicine, New York University School of Medicine, New York, New York, USA), Michael Hoffmeister (Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany), John L. Hopper (Melbourne School of Population Health, The University of Melbourne, Melbourne, Victoria, Australia), Li Hsu (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA and Department of Biostatistics, University of Washington, Seattle, Washington, USA), Thomas J. Hudson (Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada), David J. Hunter (Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA), Carolyn M. Hutter (Division of Cancer Control and Population Sciences, National Cancer Institute, US NIH, Bethesda, Maryland, USA), Rebecca D. Jackson (Division of Endocrinology, Diabetes and Metabolism, Ohio State University, Columbus, Ohio, USA), Mark A. Jenkins (Melbourne School of Population Health, The University of Melbourne, Melbourne, Victoria, Australia), Shuo Jiao (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Sébastien Küry (Service de Génétique Médicale, CHU Nantes, Nantes, France), Loic Le Marchand (Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA), Mathieu Lemire (Ontario Institute for Cancer Research, Toronto, Ontario, Canada), Noralane M. Lindor (Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona, USA), Jing Ma (Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA), Polly A. Newcomb (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA and Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA), Ulrike Peters (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA and Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA), John D. Potter (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA, Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA and Centre for Public Health Research, Massey University, Palmerston North, New Zealand), Conghui Qu (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Thomas Rohan (Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, USA), Robert E. Schoen (Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA), Fredrick R. Schumacher (Department of Preventive Medicine, University of Southern California, Los Angeles, California, USA), Daniela Seminara (Division of Cancer Control and Population Sciences, National Cancer Institute, US NIH, Bethesda, Maryland, USA), Martha L. Slattery (Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, USA), Stephen N. Thibodeau (Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA and Department of Laboratory Genetics, Mayo Clinic, Rochester, Minnesota, USA), Emily White (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA and Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington, USA) and Brent W. Zanke (Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada).

Investigators (institution and location) from the CORECT consortium include (in alphabetical order) Kendra Blalock (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Peter T. Campbell (Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA), Graham Casey (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), David V. Conti (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Christopher K. Edlund (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Jane Figueiredo (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), W. James Gauderman (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Jian Gong (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Roger C. Green (Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada), Stephen B. Gruber (University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), John F. Harju (University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA), Tabitha A. Harrison (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Eric J. Jacobs (Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA), Mark A. Jenkins (Melbourne School of Population Health, The University of Melbourne, Melbourne, Victoria, Australia), Shuo Jiao (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Li Li (Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA), Yi Lin (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Frank J. Manion (University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA), Victor Moreno (Institut d'Investigació Biomèdica de Bellvitge, Institut Catala d'Oncologia, Hospitalet, Barcelona, Spain), Bhramar Mukherjee (University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, Michigan, USA), Ulrike Peters (Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA), Leon Raskin (University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Fredrick R. Schumacher (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA), Daniela Seminara (Division of Cancer Control and Population Sciences, National Cancer Institute, US NIH, Bethesda, Maryland, USA), Gianluca Severi (Melbourne School of Population Health, The University of Melbourne, Melbourne, Victoria, Australia), Stephanie L. Stenzel (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA) and Duncan C. Thomas (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA).

The CCFR consortium is represented by Graham Casey (Department of Preventive Medicine, University of Southern California Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA).

We also thank B. Buecher of ASTERISK; U. Handte-Daub, M. Celik, R. Hettler-Jensen, U. Benscheid and U. Eilber of DACHS; and P. Soule, H. Ranu, I. Devivo, D.J. Hunter, Q. Guo, L. Zhu and H. Zhang of HPFS, NHS and PHS, as well as the following state cancer registries for their help: Alabama, Arizona, Arkansas, California, Colorado, Connecticut, Delaware, Florida, Georgia, Idaho, Illinois, Indiana, Iowa, Kentucky, Louisiana, Maine, Maryland, Massachusetts, Michigan, Nebraska, New Hampshire, New Jersey, New York, North Carolina, North Dakota, Ohio, Oklahoma, Oregon, Pennsylvania, Rhode Island, South Carolina, Tennessee, Texas, Virginia, Washington and Wyoming. We thank C. Berg and P. Prorok of PLCO; T. Riley of Information Management Services, Inc.; B. O'Brien of Westat, Inc.; B. Kopp and W. Shao of SAIC-Frederick; the WHI investigators (see https://www.whi.org/researchers/SitePages/Write%20a%20Paper.aspx) and the GECCO Coordinating Center. Participating studies (grant support) in the GECCO, CORECT and CCFR GWAS meta-analysis are GECCO (US NIH, U01CA137088 and R01CA059045), DALS (US NIH, R01CA048998), DACHS (German Federal Ministry of Education and Research, BR 1704/6-1, BR 1704/6-3, BR 1704/6-4, CH 117/1-1, 01KH0404 and 01ER0814), HPFS (US NIH, P01CA055075, UM1CA167552, R01137178 and P50CA127003), NHS (US NIH, R01137178, P50CA127003 and P01CA087969), OFCCR (US NIH, U01CA074783), PMH (US NIH, R01CA076366), PHS (US NIH, R01CA042182), VITAL (US NIH, K05CA154337), WHI (US NIH, HHSN268201100046C, HHSN268201100001C, HHSN268201100002C, HHSN268201100003C, HHSN268201100004C, HHSN271201100004C and 268200764316C) and PLCO (US NIH, Z01CP 010200, U01HG004446 and U01HG 004438). CORECT is supported by the National Cancer Institute as part of the GAME-ON consortium (US NIH, U19CA148107) with additional support from National Cancer Institute grants (R01CA81488 and P30CA014089), the National Human Genome Research Institute at the US NIH (T32HG000040) and the National Institute of Environmental Health Sciences at the US NIH (T32ES013678). CCFR is supported by the National Cancer Institute, US NIH under RFA CA-95-011 and through cooperative agreements with members of the Colon Cancer Family Registry and principal investigators of the Australasian Colorectal Cancer Family Registry (US NIH, U01CA097735), the Familial Colorectal Neoplasia Collaborative Group (US NIH, U01CA074799) (University of Southern California), the Mayo Clinic Cooperative Family Registry for Colon Cancer Studies (US NIH, U01CA074800), the Ontario Registry for Studies of Familial Colorectal Cancer (US NIH, U01CA074783), the Seattle Colorectal Cancer Family Registry (US NIH, U01CA074794) and the University of Hawaii Colorectal Cancer Family Registry (US NIH, U01CA074806). The GWAS work was supported by a National Cancer Institute grant (US NIH, U01CA122839). OFCCR was supported by a GL2 grant from the Ontario Research Fund, Canadian Institutes of Health Research and a Cancer Risk Evaluation (CaRE) Program grant from the Canadian Cancer Society Research Institute. T.J. Hudson and B.W. Zanke are recipients of Senior Investigator Awards from the Ontario Institute for Cancer Research, through support from the Ontario Ministry of Economic Development and Innovation. ASTERISK was funded by a Regional Hospital Clinical Research Program (PHRC) and supported by the Regional Council of Pays de la Loire, the Groupement des Entreprises Françaises dans la Lutte contre le Cancer (GEFLUC), the Association Anne de Bretagne Génétique and the Ligue Régionale Contre le Cancer (LRCC). PLCO data sets were accessed with approval through dbGaP (CGEMS prostate cancer scan, phs000207.v1.p1; CGEMS pancreatic cancer scan, phs000206.v4.p3; and GWAS of Lung Cancer and Smoking, phs000093.v2.p2, which was funded by Z01CP 010200, U01HG004446 and U01HG 004438 from the US NIH).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

W.Z. conceived and directed the Asia Colorectal Cancer Consortium and the Shanghai-Vanderbilt Colorectal Cancer Genetics Project. W.-H.J. and Y.-X.Z.; K. Matsuda; S.-S.K.; K. Matsuo; X.-O.S., Y.-B.X. and Y.-T.G.; A.S.; S.H.J.; and D.-H.K. directed CRC projects for the Guangzhou Colorectal Cancer Study, the BioBank Japan Colorectal Cancer Study, the Hwasun Cancer Epidemiology Study–Colon and Rectum Cancer (HCES-CRC), the Aichi Colorectal Cancer Study, the Shanghai studies, the Korea-NCC (National Cancer Center) Colorectal Cancer Study, the KCPS-II Colorectal Cancer Study and the Korea-Seoul Colorectal Cancer Study, respectively. B.Z., Q.C. and W.W. coordinated the project. Q.C. directed laboratory operations. J.S. performed the genotyping experiments. B.Z. performed the statistical and bioinformatics analyses. W.W. contributed to the statistical analyses and data interpretation. A.T. conducted the statistical analyses and imputation for BioBank Japan. B.Z., W.W. and J.L. managed the data. Y.Z. and B.Z. performed the expression analysis for TCGA data. B.Z. and W.Z. wrote the manuscript with significant contributions from X.-O.S., Q.C., J.L., W.W., B.L. and Y.Z. All authors contributed to data and biological sample collection in the original studies included in this project and to manuscript revision. All authors have reviewed and approved the content of the paper.

Corresponding author

Correspondence to Wei Zheng.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A complete list of members and affiliations appears in the Acknowledgments.

A complete list of members and affiliations appears in the Acknowledgments.

A complete list of members and affiliations appears in the Acknowledgments.

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Tables 1–20 and Supplementary Figures 1–6 (PDF 9786 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, B., Jia, WH., Matsuda, K. et al. Large-scale genetic study in East Asians identifies six new loci associated with colorectal cancer risk. Nat Genet 46, 533–542 (2014). https://doi.org/10.1038/ng.2985

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2985

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing